1
|
Chen P, Wang S, Zhang H, Li J. Recent advances in nanotherapy-based treatment of epilepsy. Colloids Surf B Biointerfaces 2025; 249:114499. [PMID: 39778465 DOI: 10.1016/j.colsurfb.2025.114499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/21/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures affecting millions of people worldwide. Despite advances in drug therapy, a significant proportion of patients remain resistant to conventional antiepileptic drugs (AEDs) due to challenges such as impermeability of the blood-brain barrier (BBB), multidrug resistance, and multifaceted epileptogenesis. Nanotechnology offers promising strategies to overcome these barriers by enhancing drug delivery across the BBB, improving target specificity and minimizing systemic side effects. This review explores recent advances in different innovative strategies of nanodelivery systems for epilepsy therapy, and we will discuss the design principles, mechanisms of action and therapeutic efficacy of these nanodelivery systems. In addition, we discuss the challenges and limitations that hinder the clinical translation of nanomedicine-based therapies for epilepsy. We emphasize the need for personalized and multidisciplinary approaches as well as the importance of continued research and interdisciplinary collaboration in order to translate these innovative strategies into effective therapies. Ultimately, the use of nanotechnology has the potential to enhance seizure control, reduce the burden of epilepsy, and improve the quality of life of patients affected by this complex neurological disorder. Nanotechnology-based drug delivery systems may usher in a new era of precision medicine for epilepsy treatment.
Collapse
Affiliation(s)
- Peng Chen
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Shudong Wang
- Jinzhou Medical University, Liaoning 121001, China
| | - Heming Zhang
- Dalian Medical University, Liaoning 116044, China
| | - Jian Li
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
2
|
Perucca E, Taglialatela M. Targeting Kv7 Potassium Channels for Epilepsy. CNS Drugs 2025; 39:263-288. [PMID: 39853501 PMCID: PMC11850491 DOI: 10.1007/s40263-024-01155-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2024] [Indexed: 01/26/2025]
Abstract
Voltage-gated Kv7 potassium channels, particularly Kv7.2 and Kv.7.3 channels, play a critical role in modulating susceptibility to seizures, and mutations in genes that encode these channels cause heterogeneous epilepsy phenotypes. On the basis of this evidence, activation of Kv7.2 and Kv.7.3 channels has long been considered an attractive target in the search for novel antiseizure medications. Ezogabine (retigabine), the first Kv7.2/3 activator introduced in 2011 for the treatment of focal seizures, was withdrawn from the market in 2017 due to declining use after discovery of its association with pigmentation changes in the retina, skin, and mucosae. A novel formulation of ezogabine for pediatric use (XEN496) has been recently investigated in children with KCNQ2-related developmental and epileptic encephalopathy, but the trial was terminated prematurely for reasons unrelated to safety. Among novel Kv7.2/3 openers in clinical development, azetukalner has shown dose-dependent efficacy against drug-resistant focal seizures with a good tolerability profile and no evidence of pigmentation-related adverse effects in early clinical studies, and it is now under investigation in phase III trials for the treatment of focal seizures, generalized tonic-clonic seizures, and major depressive disorder. Another Kv7.2/3 activator, BHV-7000, has completed phase I studies in healthy subjects, with excellent tolerability at plasma drug concentrations that exceed the median effective concentrations in a preclinical model of anticonvulsant activity, but no efficacy data in patients with epilepsy are available to date. Among other Kv7.2/3 activators in clinical development as potential antiseizure medications, pynegabine and CB-003 have completed phase I safety and pharmacokinetic studies, but results have not been yet reported. Overall, interest in targeting Kv7 channels for the treatment of epilepsy and for other indications remains strong. Future breakthroughs in this area could come from exploitation of mechanistic differences in the action of Kv7 activators, and from the development of molecules that combine Kv7 activation with other mechanisms of action.
Collapse
Affiliation(s)
- Emilio Perucca
- Department of Medicine (Austin Health), Melbourne Brain Center, The University of Melbourne, 245 Burgundy St., Heidelberg, VIC, 3084, Australia.
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
3
|
Lu TL, Liutkevičienė R, Rovite V, Gao ZH, Wu SN. Evaluation of Small-Molecule Candidates as Modulators of M-Type K + Currents: Impacts on Current Amplitude, Gating, and Voltage-Dependent Hysteresis. Int J Mol Sci 2025; 26:1504. [PMID: 40003973 PMCID: PMC11855363 DOI: 10.3390/ijms26041504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The core subunits of the KV7.2, KV7.3, and KV7.5 channels, encoded by the KCNQ2, KCNQ3, and KCNQ5 genes, are expressed across various cell types and play a key role in generating the M-type K+ current (IK(M)). This current is characterized by an activation threshold at low voltages and displays slow activation and deactivation kinetics. Variations in the amplitude and gating kinetics of IK(M) can significantly influence membrane excitability. Notably, IK(M) demonstrates distinct voltage-dependent hysteresis when subjected to prolonged isosceles-triangular ramp pulses. In this review, we explore various small-molecule modulators that can either inhibit or enhance the amplitude of IK(M), along with their perturbations on its gating kinetics and voltage-dependent hysteresis. The inhibitors of IK(M) highlighted here include bisoprolol, brivaracetam, cannabidiol, nalbuphine, phenobarbital, and remdesivir. Conversely, compounds such as flupirtine, kynurenic acid, naringenin, QO-58, and solifenacin have been shown to enhance IK(M). These modulators show potential as pharmacological or therapeutic strategies for treating certain disorders linked to gain-of-function or loss-of-function mutations in M-type K+ (KV7x or KCNQx) channels.
Collapse
Affiliation(s)
- Te-Ling Lu
- Department of Pharmacy, China Medical University, Taichung 406040, Taiwan;
| | - Rasa Liutkevičienė
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu 2, 50161 Kaunas, Lithuania;
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre (BMC), LV-1067 Riga, Latvia;
| | - Zi-Han Gao
- Institute of Basic Medical Sciences, College of Medical, National Cheng Kung University, Tainan City 701401, Taiwan;
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, College of Medical, National Cheng Kung University, Tainan City 701401, Taiwan;
- Department of Research and Education, An Nan Hospital, China Medical University, Tainan City 709204, Taiwan
- School of Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
4
|
Mao M, Jancovski N, Kushner Y, Teasdale L, Truong P, Zhou K, Reid S, Jia L, Aung YH, Li M, Reid CA, Byars S, Scheffer I, Petrou S, Maljevic S. Developmental dysfunction in a preclinical model of Kcnq2 developmental and epileptic encephalopathy. Neurobiol Dis 2025; 205:106782. [PMID: 39733957 DOI: 10.1016/j.nbd.2024.106782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/23/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Developmental and epileptic encephalopathies (DEE) are rare but severe neurodevelopmental disorders characterised by early-onset seizures often combined with developmental delay, behavioural and cognitive deficits. Treatment for DEEs is currently limited to seizure control and provides no benefits to the patients' developmental and cognitive outcomes. Genetic variants are the most common cause of DEE with KCNQ2 being one of the most frequently identified disease-causing genes. KCNQ2 encodes a voltage-gated potassium channel KV7.2 widely expressed in the central nervous system and critically involved in the regulation of neuronal excitability. In this study, we aimed to characterise a KCNQ2 variant (K556E) found in a female patient with DEE using a heterologous expression system and a knock-in mouse model. METHODS Wild-type KCNQ2 or K556E variant were expressed in Chinese Hamster Ovary (CHO) cells (with or without KCNQ3) and their biophysical properties assessed using patch clamp recordings. We further engineered a new Kcnq2 DEE mouse model (K557E) based on the K556E variant and characterised it using behavioural, electrophysiological, and transcriptome analysis. RESULTS A mild loss of function was observed only when the mutant channel was co-expressed with KCNQ3 in the heterologous system. The heterozygous knock-in mice showed a reduced survival rate and increased susceptibility to induced seizures. Electrophysiology recordings in brain slices revealed a hyperexcitable phenotype for cortical layer 2/3 pyramidal neurons with retigabine (KV7 channel opener) able to rescue both the increased sensitivity to chemically-induced seizures in vivo and neuronal excitability ex vivo. Whole-brain RNA sequencing revealed numerous differentially expressed genes and biological pathways pointing at dysregulation of early developmental processes. CONCLUSIONS Our study reports on a novel Kcnq2 DEE mouse model recapitulating aspects of the disease phenotype with the electrophysiological and transcriptome analysis providing insights into KCNQ2 DEE mechanisms that can be leveraged for future therapy development.
Collapse
Affiliation(s)
- Miaomiao Mao
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Nikola Jancovski
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Yafit Kushner
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Lucas Teasdale
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Phan Truong
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Kun Zhou
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Samuel Reid
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Linghan Jia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Ye Htet Aung
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Melody Li
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Sean Byars
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Ingrid Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia; Praxis Precision Medicine Inc, Cambridge, MA 02142, USA.
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.
| |
Collapse
|
5
|
Filareto I, Mosca I, Freri E, Ragona F, Canafoglia L, Solazzi R, Castellotti B, Messina G, Gellera C, Soldovieri MV, Ambrosino P, Taglialatela M, DiFrancesco JC, Granata T. Pharmacological approaches in drug-resistant pediatric epilepsies caused by pathogenic variants in potassium channel genes. Front Cell Neurosci 2025; 18:1512365. [PMID: 39926415 PMCID: PMC11802495 DOI: 10.3389/fncel.2024.1512365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
Variants in genes encoding for voltage-gated K+ (Kv) channels are frequent cause of drug-resistant pediatric epilepsies. Obtaining a molecular diagnosis gives the opportunity to assess the efficacy of pharmacological strategies based on in vitro features of mutant channels. In this retrospective observational study, we selected patients with drug-resistant pediatric epilepsies caused by variants in potassium channel encoding genes, followed at the Fondazione IRCCS Istituto Neurologico Carlo Besta of Milan, Italy. After the experimental characterization of variants' functional properties in transiently transfected Chinese Hamster Ovary (CHO) cells, we identified drugs to be used as pharmacological approaches. We recruited six patients carrying different missense variants in four Kv channels (Kv7.2, Kv7.3, Kv3.1, and KNa1.1). In vitro experiments demonstrated that variants in Kv7 channels induced loss-of-function (LoF) effects, while those affecting Kv3.1 or KNa1.1 led to gain-of-function (GoF). Moreover, we found that the Kv7 channels activator gabapentin was able to revert the LoF effects caused by Kv7.2/Kv7.3 variants, and the potassium channel-blocker fluoxetine counteracted the GoF effects in Kv3.1 or KNa1.1 variants. According to experimental data, patients carrying Kv7 variants were treated with gabapentin. While this treatment resulted successful in two patients (#1, Kv7.2 G310S variant; #3, Kv7.3 V359L + Kv7.3 D542N), it resulted detrimental in the remaining case (#2, Kv7.2 D535E), requiring drug withdrawal. The application in vivo of fluoxetine to counteract GoF effects induced by Kv3.1 or KNa1.1 variants determined a significant reduction of both seizure frequency and behavior disturbances in patient #4 (Kv3.1 V425M), and in both subjects carrying KNa1.1 variants (#5, S937G and #6, R262Q). However, for the latter case, this drug was halted due to severe behavioral side effects. For most of the patients herein reported, pharmacological strategies, selected according to the in vitro functional properties of Kv-channels pathogenic variants, resulted in a significant improvement of both epileptic and cognitive features.
Collapse
Affiliation(s)
- Ilaria Filareto
- Department of Medical and Surgical Sciences of the Mothers, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Ilaria Mosca
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Elena Freri
- Department of Pediatric Neuroscience, member of the European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Ragona
- Department of Pediatric Neuroscience, member of the European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Canafoglia
- Department of Epileptology, member of the European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Roberta Solazzi
- Department of Pediatric Neuroscience, member of the European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuliana Messina
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Virginia Soldovieri
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Paolo Ambrosino
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | | | | | - Tiziana Granata
- Department of Pediatric Neuroscience, member of the European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
6
|
Nappi M, Alberini G, Berselli A, Roscioni A, Soldovieri MV, Servettini I, Barrese V, Weckhuysen S, Chiu TGA, Scheffer IE, Benfenati F, Maragliano L, Miceli F, Taglialatela M. Constitutive opening of the Kv7.2 pore activation gate causes KCNQ2-developmental encephalopathy. Proc Natl Acad Sci U S A 2024; 121:e2412388121. [PMID: 39602259 PMCID: PMC11626135 DOI: 10.1073/pnas.2412388121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Pathogenic variants in KCNQ2 encoding Kv7.2 voltage-gated potassium channel subunits cause developmental encephalopathies (KCNQ2-encephalopathies), both with and without epilepsy. We herein describe the clinical, in vitro, and in silico features of two encephalopathy-causing variants (A317T, L318V) in Kv7.2 affecting two consecutive residues in the S6 activation gate that undergoes large structural rearrangements during pore opening; the disease-causing A356T variant in KCNQ3, paralogous to the A317T variant in KCNQ2, was also investigated. Currents through KCNQ2 mutant channels displayed increased density, hyperpolarizing shifts in activation gating, faster activation and slower deactivation kinetics, and resistance to changes in the cellular concentrations of phosphatidylinositol 4,5-bisphosphate (PIP2), a critical regulator of Kv7 channel function; all these features are consistent with a strong gain-of-function effect. An increase in the probability of single-channel opening, with no change in membrane abundance or single-channel conductance, was responsible for the observed gain-of-function effects. All-atom molecular dynamics simulations revealed that the mutations widened the inner pore gate and stabilized a constitutively open channel configuration in the closed state, with minimal effects on the open conformation. Thus, mutation-induced stabilization of the inner pore gate open configuration is a molecular pathogenetic mechanism for KCNQ2-related encephalopathies.
Collapse
Affiliation(s)
- Mario Nappi
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Giulio Alberini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova16132, Italy
| | - Alessandro Berselli
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Experimental Medicine, Università degli Studi di Genova, Genova16132, Italy
| | - Agnese Roscioni
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona60131, Italy
| | | | - Ilenio Servettini
- Department of Medicine and Health Science, University of Molise, Campobasso86100, Italy
| | - Vincenzo Barrese
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, Antwerp2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp2610, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp2610, Belgium
| | - Ting-Gee Annie Chiu
- Division of Medicine, Dentistry and Health Sciences, University of Melbourne, Austin Health, Melbourne, VIC3084, Australia
| | - Ingrid E. Scheffer
- The Florey Institute of Neuroscience and Mental Health and Murdoch Children’s Research Institutes, University of Melbourne, Austin and Royal Children’s Hospital, Melbourne, VIC3052, Australia
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova16132, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova16132, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona60131, Italy
| | - Francesco Miceli
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| | - Maurizio Taglialatela
- Department of Neuroscience, Section of Pharmacology, University of Naples Federico II, Naples80131, Italy
| |
Collapse
|
7
|
Abbott GW, Manville RW. Discovery of a potent, Kv7.3-selective potassium channel opener from a Polynesian traditional botanical anticonvulsant. Commun Chem 2024; 7:233. [PMID: 39390220 PMCID: PMC11467302 DOI: 10.1038/s42004-024-01318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Plants remain an important source of biologically active small molecules with high therapeutic potential. The voltage-gated potassium (Kv) channel formed by Kv7.2/3 (KCNQ2/3) heteromers is a major target for anticonvulsant drug development. Here, we screened 1444 extracts primarily from plants collected in California and the US Virgin Islands, for their ability to activate Kv7.2/3 but not inhibit Kv1.3, to select against tannic acid being the active component. We validated the 7 strongest hits, identified Thespesia populnea (miro, milo, portia tree) as the most promising, then discovered its primary active metabolite to be gentisic acid (GA). GA highly potently activated Kv7.2/3 (EC50, 2.8 nM). GA is, uniquely to our knowledge, 100% selective for Kv7.3 versus other Kv7 homomers; it requires S5 residue Kv7.3-W265 for Kv7.2/3 activation, and it ameliorates pentylenetetrazole-induced seizures in mice. Structure-activity studies revealed that the FDA-approved vasoprotective drug calcium dobesilate, a GA analog, is a previously unrecognized Kv7.2/3 channel opener. Also an active aspirin metabolite, GA provides a molecular rationale for the use of T. populnea as an anticonvulsant in Polynesian indigenous medicine and presents novel pharmacological prospects for potent, isoform-selective, therapeutic Kv7 channel activation.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
8
|
Edmond MA, Hinojo-Perez A, Efrem M, Yi-Chun L, Shams I, Hayoz S, de la Cruz A, Perez Rodriguez ME, Diaz-Solares M, Dykxhoorn DM, Luo YL, Barro-Soria R. Lipophilic compounds restore function to neurodevelopmental-associated KCNQ3 mutations. Commun Biol 2024; 7:1181. [PMID: 39300259 DOI: 10.1038/s42003-024-06873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
A major driver of neuronal hyperexcitability is dysfunction of K+ channels, including voltage-gated KCNQ2/3 channels. Their hyperpolarized midpoint of activation and slow activation and deactivation kinetics produce a current that regulates membrane potential and impedes repetitive firing. Inherited mutations in KCNQ2 and KCNQ3 are linked to a wide spectrum of neurodevelopmental disorders (NDDs), ranging from benign familial neonatal seizures to severe epileptic encephalopathies and autism spectrum disorders. However, the impact of these variants on the molecular mechanisms underlying KCNQ3 channel function remains poorly understood and existing treatments have significant side effects. Here, we use voltage clamp fluorometry, molecular dynamic simulations, and electrophysiology to investigate NDD-associated variants in KCNQ3 channels. We identified two distinctive mechanisms by which loss- and gain-of function NDD-associated mutations in KCNQ3 affect channel gating: one directly affects S4 movement while the other changes S4-to-pore coupling. MD simulations and electrophysiology revealed that polyunsaturated fatty acids (PUFAs) primarily target the voltage-sensing domain in its activated conformation and form a weaker interaction with the channel's pore. Consistently, two such compounds yielded partial and complete functional restoration in R227Q- and R236C-containing channels, respectively. Our results reveal the potential of PUFAs to be developed into therapies for diverse KCNQ3-based channelopathies.
Collapse
Affiliation(s)
- Michaela A Edmond
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Texas A&M University Health Science Center, Department of Neuroscience & Experimental Therapeutics, Bryan, USA
| | - Andy Hinojo-Perez
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mekedlawit Efrem
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Lin Yi-Chun
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Iqra Shams
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sebastien Hayoz
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Physiology, University of Arizona, Tucson, USA
| | - Alicia de la Cruz
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
- Linkoping University, Department of Biomedical and Clinical Sciences (BKV), Linkoping, Sweden
| | | | - Maykelis Diaz-Solares
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Yun Lyna Luo
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Rene Barro-Soria
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
9
|
Ray S, Stampf JL, Kudlacek O, Yang JW, Schicker KW, Graf Y, Losgott T, Boehm S, Salzer I. A triple cysteine motif as major determinant of the modulation of neuronal K V7 channels by the paracetamol metabolite N-acetyl-p-benzo quinone imine. Br J Pharmacol 2024; 181:2851-2868. [PMID: 38657956 DOI: 10.1111/bph.16380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/19/2024] [Accepted: 03/10/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND AND PURPOSE The analgesic action of paracetamol involves KV7 channels, and its metabolite N-acetyl-p-benzo quinone imine (NAPQI), a cysteine modifying reagent, was shown to increase currents through such channels in nociceptors. Modification of cysteine residues by N-ethylmaleimide, H2O2, or nitric oxide has been found to modulate currents through KV7 channels. The study aims to identify whether, and if so which, cysteine residues in neuronal KV7 channels might be responsible for the effects of NAPQI. EXPERIMENTAL APPROACH To address this question, we used a combination of perforated patch-clamp recordings, site-directed mutagenesis, and mass spectrometry applied to recombinant KV7.1 to KV7.5 channels. KEY RESULTS Currents through the cardiac subtype KV7.1 were reduced by NAPQI. Currents through all other subtypes were increased, either by an isolated shift of the channel voltage dependence to more negative values (KV7.3) or by such a shift combined with increased maximal current levels (KV7.2, KV7.4, KV7.5). A stretch of three cysteine residues in the S2-S3 linker region of KV7.2 was necessary and sufficient to mediate these effects. CONCLUSION AND IMPLICATION The paracetamol metabolite N-acetyl-p-benzo quinone imine (NAPQI) modifies cysteine residues of KV7 subunits and reinforces channel gating in homomeric and heteromeric KV7.2 to KV7.5, but not in KV7.1 channels. In KV7.2, a triple cysteine motif located within the S2-S3 linker region mediates this reinforcement that can be expected to reduce the excitability of nociceptors and to mediate antinociceptive actions of paracetamol.
Collapse
Affiliation(s)
- Sutirtha Ray
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jan-Luca Stampf
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus W Schicker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Yvonne Graf
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Losgott
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Di Matteo F, Mancuso F, Turcio R, Ciaglia T, Stagno C, Di Chio C, Campiglia P, Bertamino A, Giofrè SV, Ostacolo C, Iraci N. KCNT1 Channel Blockers: A Medicinal Chemistry Perspective. Molecules 2024; 29:2940. [PMID: 38931004 PMCID: PMC11206332 DOI: 10.3390/molecules29122940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Potassium channels have recently emerged as suitable target for the treatment of epileptic diseases. Among potassium channels, KCNT1 channels are the most widely characterized as responsible for several epileptic and developmental encephalopathies. Nevertheless, the medicinal chemistry of KCNT1 blockers is underdeveloped so far. In the present review, we describe and analyse the papers addressing the issue of KCNT1 blockers' development and identification, also evidencing the pros and the cons of the scientific approaches therein described. After a short introduction describing the epileptic diseases and the structure-function of potassium channels, we provide an extensive overview of the chemotypes described so far as KCNT1 blockers, and the scientific approaches used for their identification.
Collapse
Affiliation(s)
- Francesca Di Matteo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Francesca Mancuso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Rita Turcio
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Claudio Stagno
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Carla Di Chio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Salvatore Vincenzo Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy (R.T.); (T.C.)
| | - Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
11
|
Iraci N, Carotenuto L, Ciaglia T, Belperio G, Di Matteo F, Mosca I, Carleo G, Giovanna Basilicata M, Ambrosino P, Turcio R, Puzo D, Pepe G, Gomez-Monterrey I, Soldovieri MV, Di Sarno V, Campiglia P, Miceli F, Bertamino A, Ostacolo C, Taglialatela M. In Silico Assisted Identification, Synthesis, and In Vitro Pharmacological Characterization of Potent and Selective Blockers of the Epilepsy-Associated KCNT1 Channel. J Med Chem 2024; 67:9124-9149. [PMID: 38782404 PMCID: PMC11181338 DOI: 10.1021/acs.jmedchem.4c00268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Gain-of-function (GoF) variants in KCNT1 channels cause severe, drug-resistant forms of epilepsy. Quinidine is a known KCNT1 blocker, but its clinical use is limited due to severe drawbacks. To identify novel KCNT1 blockers, a homology model of human KCNT1 was built and used to screen an in-house library of compounds. Among the 20 molecules selected, five (CPK4, 13, 16, 18, and 20) showed strong KCNT1-blocking ability in an in vitro fluorescence-based assay. Patch-clamp experiments confirmed a higher KCNT1-blocking potency of these compounds when compared to quinidine, and their selectivity for KCNT1 over hERG and Kv7.2 channels. Among identified molecules, CPK20 displayed the highest metabolic stability; this compound also blocked KCNT2 currents, although with a lower potency, and counteracted GoF effects prompted by 2 recurrent epilepsy-causing KCNT1 variants (G288S and A934T). The present results provide solid rational basis for future design of novel compounds to counteract KCNT1-related neurological disorders.
Collapse
Affiliation(s)
- Nunzio Iraci
- Department
of Chemical, Biological, Pharmaceutical and Environmental Sciences
(CHIBIOFARAM), University of Messina, Viale F. Stagno d’Alcontres
31, 98166 Messina, Italy
| | - Lidia Carotenuto
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Tania Ciaglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Giorgio Belperio
- Department
of Science and Technology, University of
Sannio, Via F. De Sanctis, 82100 Benevento, Italy
| | - Francesca Di Matteo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Ilaria Mosca
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Giusy Carleo
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Manuela Giovanna Basilicata
- Department
of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, P.zza L. Miraglia 2, 80138 Naples, Italy
| | - Paolo Ambrosino
- Department
of Science and Technology, University of
Sannio, Via F. De Sanctis, 82100 Benevento, Italy
| | - Rita Turcio
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Deborah Puzo
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Giacomo Pepe
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Isabel Gomez-Monterrey
- Department
of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Maria Virginia Soldovieri
- Department
of Medicine and Health Science Vincenzo Tiberio, University of Molise, Via C. Gazzani, 86100 Campobasso, Italy
| | - Veronica Di Sarno
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Pietro Campiglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Francesco Miceli
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Alessia Bertamino
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - Maurizio Taglialatela
- Department
of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
12
|
Graziano B, Wang L, White OR, Kaplan DH, Fernandez-Abascal J, Bianchi L. Glial KCNQ K + channels control neuronal output by regulating GABA release from glia in C. elegans. Neuron 2024; 112:1832-1847.e7. [PMID: 38460523 PMCID: PMC11156561 DOI: 10.1016/j.neuron.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/11/2024]
Abstract
KCNQs are voltage-gated K+ channels that control neuronal excitability and are mutated in epilepsy and autism spectrum disorder (ASD). KCNQs have been extensively studied in neurons, but their function in glia is unknown. Using voltage, calcium, and GABA imaging, optogenetics, and behavioral assays, we show here for the first time in Caenorhabditis elegans (C. elegans) that glial KCNQ channels control neuronal excitability by mediating GABA release from glia via regulation of the function of L-type voltage-gated Ca2+ channels. Further, we show that human KCNQ channels have the same role when expressed in nematode glia, underscoring conservation of function across species. Finally, we show that pathogenic loss-of-function and gain-of-function human KCNQ2 mutations alter glia-to-neuron GABA signaling in distinct ways and that the KCNQ channel opener retigabine exerts rescuing effects. This work identifies glial KCNQ channels as key regulators of neuronal excitability via control of GABA release from glia.
Collapse
Affiliation(s)
- Bianca Graziano
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lei Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Olivia R White
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daryn H Kaplan
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
13
|
Stott JB, Greenwood IA. G protein βγ regulation of KCNQ-encoded voltage-dependent K channels. Front Physiol 2024; 15:1382904. [PMID: 38655029 PMCID: PMC11035767 DOI: 10.3389/fphys.2024.1382904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The KCNQ family is comprised of five genes and the expression products form voltage-gated potassium channels (Kv7.1-7.5) that have a major impact upon cellular physiology in many cell types. Each functional Kv7 channel forms as a tetramer that often associates with proteins encoded by the KCNE gene family (KCNE1-5) and is critically reliant upon binding of phosphatidylinositol bisphosphate (PIP2) and calmodulin. Other modulators like A-kinase anchoring proteins, ubiquitin ligases and Ca-calmodulin kinase II alter Kv7 channel function and trafficking in an isoform specific manner. It has now been identified that for Kv7.4, G protein βγ subunits (Gβγ) can be added to the list of key regulators and is paramount for channel activity. This article provides an overview of this nascent field of research, highlighting themes and directions for future study.
Collapse
Affiliation(s)
| | - Iain A. Greenwood
- Vascular Biology Research Group, Institute of Molecular and Clinical Sciences, St George’s University of London, London, United Kingdom
| |
Collapse
|
14
|
Sun H, Undem BJ. Selective KCNQ2/3 Potassium Channel Opener ICA-069673 Inhibits Excitability in Mouse Vagal Sensory Neurons. J Pharmacol Exp Ther 2024; 389:118-127. [PMID: 38290975 PMCID: PMC10949160 DOI: 10.1124/jpet.123.001959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 01/18/2024] [Indexed: 02/01/2024] Open
Abstract
Heightened excitability of vagal sensory neurons in inflammatory visceral diseases contributes to unproductive and difficult-to-treat neuronally based symptoms such as visceral pain and dysfunction. Identification of targets and modulators capable of regulating the excitability of vagal sensory neurons may lead to novel therapeutic options. KCNQ1-KCNQ5 genes encode KV7.1-7.5 potassium channel α-subunits. Homotetrameric or heterotetrameric KV7.2-7.5 channels can generate the so-called M-current (IM) known to decrease the excitability of neurons including visceral sensory neurons. This study aimed to address the hypothesis that KV7.2/7.3 channels are key regulators of vagal sensory neuron excitability by evaluating the effects of KCNQ2/3-selective activator, ICA-069673, on IM in mouse nodose neurons and determining its effects on excitability and action potential firings using patch clamp technique. The results showed that ICA-069673 enhanced IM density, accelerated the activation, and delayed the deactivation of M-channels in a concentration-dependent manner. ICA-069673 negatively shifted the voltage-dependent activation of IM and increased the maximal conductance. Consistent with its effects on IM, ICA-069673 induced a marked hyperpolarization of resting potential and reduced the input resistance. The hyperpolarizing effect was more pronounced in partially depolarized neurons. Moreover, ICA-069673 caused a 3-fold increase in the minimal amount of depolarizing current needed to evoke an action potential, and significantly limited the action potential firings in response to sustained suprathreshold stimulations. ICA-069673 had no effect on membrane currents when Kcnq2 and Kcnq3 were deleted. These results indicate that opening KCNQ2/3-mediated M-channels is sufficient to suppress the excitability and enhance spike accommodation in vagal visceral sensory neurons. SIGNIFICANCE STATEMENT: This study supports the hypothesis that selectively activating KCNQ2/3-mediated M-channels is sufficient to suppress the excitability and action potential firings in vagal sensory neurons. These results provide evidence in support of further investigations into the treatment of various visceral disorders that involve nociceptor hyperexcitability with selective KCNQ2/3 M-channel openers.
Collapse
Affiliation(s)
- Hui Sun
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bradley J Undem
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
15
|
Celentano C, Carotenuto L, Miceli F, Carleo G, Corrado B, Baroli G, Iervolino S, Vecchione R, Taglialatela M, Barrese V. Kv7 channel activation reduces brain endothelial cell permeability and prevents kainic acid-induced blood-brain barrier damage. Am J Physiol Cell Physiol 2024; 326:C893-C904. [PMID: 38284124 PMCID: PMC11193483 DOI: 10.1152/ajpcell.00709.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Ion channels in the blood-brain barrier (BBB) play a main role in controlling the interstitial fluid composition and cerebral blood flow, and their dysfunction contributes to the disruption of the BBB occurring in many neurological diseases such as epilepsy. In this study, using morphological and functional approaches, we evaluated the expression and role in the BBB of Kv7 channels, a family of voltage-gated potassium channels including five members (Kv7.1-5) that play a major role in the regulation of cell excitability and transmembrane flux of potassium ions. Immunofluorescence experiments showed that Kv7.1, Kv7.4, and Kv7.5 were expressed in rat brain microvessels (BMVs), as well as brain primary- and clonal (BEND-3) endothelial cells (ECs). Kv7.5 localized at the cell-to-cell junction sites, whereas Kv7.4 was also found in pericytes. The Kv7 activator retigabine increased transendothelial electrical resistance (TEER) in both primary ECs and BEND-3 cells; moreover, retigabine reduced paracellular dextran flux in BEND-3 cells. These effects were prevented by the selective Kv7 blocker XE-991. Exposure to retigabine also hyperpolarized cell membrane and increased tight junctions (TJs) integrity in BEND-3 cells. BMVs from rats treated with kainic acid (KA) showed a disruption of TJs and a selective reduction of Kv7.5 expression. In BEND-3 cells, retigabine prevented the increase of cell permeability and the reduction of TJs integrity induced by KA. Overall, these findings demonstrate that Kv7 channels are expressed in the BBB, where they modulate barrier properties both in physiological and pathological conditions.NEW & NOTEWORTHY This study describes for the first time the expression and the functional role of Kv7 potassium channels in the blood-brain barrier. We show that the opening of Kv7 channels reduces endothelial cell permeability both in physiological and pathological conditions via the hyperpolarization of cell membrane and the sealing of tight junctions. Therefore, activation of endothelial Kv7 channels might be a useful strategy to treat epilepsy and other neurological disorders characterized by blood-brain barrier dysfunction.
Collapse
Affiliation(s)
- Camilla Celentano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Lidia Carotenuto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Francesco Miceli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Giusy Carleo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Brunella Corrado
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Health Care, Istituto Italiano di Tecnologia, Naples, Italy
| | - Giulia Baroli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Stefania Iervolino
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Raffaele Vecchione
- Center for Advanced Biomaterials for Health Care, Istituto Italiano di Tecnologia, Naples, Italy
| | - Maurizio Taglialatela
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Vincenzo Barrese
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| |
Collapse
|
16
|
Zhang Y, Xue Y, Ma Y, Du X, Lu B, Wang Y, Yan Z. Improved classification and pathogenicity assessment by comprehensive functional studies in a large data set of KCNQ2 variants. Life Sci 2024; 339:122378. [PMID: 38142737 DOI: 10.1016/j.lfs.2023.122378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
AIMS The paucity of functional annotations on hundreds of KCNQ2 variants impedes the diagnosis and treatment of KCNQ2-related disorders. The aims of this work were to determine the functional properties of 331 clinical KCNQ2 variants, interpreted the pathogenicity of 331 variants using functional data,and explored the association between homomeric channel functions and phenotypes. MAIN METHODS We collected 145 KCNQ2 variants from 232 epilepsy patients and 186 KCNQ2 missense variants from the ClinVar database. Whole-cell patch-clamp recording was used to classify the function of 331 variants. Subsequently, we proposed 24 criteria for the pathogenicity interpretation of KCNQ2 variants and used them to assess pathogenicity of 331 variants. Finally, we analyzed the clinical phenotypes of patients carrying these variants, and explored the correlations between functional mechanisms and phenotypes. KEY FINDINGS In the homozygous state, 287 were classified as loss-of-function and 14 as gain-of-function. In the more clinically relative heterozygous state, 200 variants exhibited functional impairment, 121 of which showed dominant-negative effects on wild-type KCNQ2 subunits. After introducing functional data as strong-level evidence to interpret pathogenicity, over half of variants (169/331) were reclassified and 254 were classified as pathogenic/likely pathogenic. Moreover, dominant-negative effect and haploinsufficiency were identified as primary mechanisms in DEE/ID and SeLNE, respectively. The degree of impairment of channel function correlated with the phenotype severity. SIGNIFICANCE Our study reveals the possible cause of KCNQ2-related disorders at the molecular level, provides compelling evidence for clinical classification of KCNQ2 variants, and expands the knowledge of correlations between functional mechanisms and phenotypes.
Collapse
Affiliation(s)
- Yuwei Zhang
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuqing Xue
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yu Ma
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaonan Du
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Zhiqiang Yan
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200438, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
17
|
Deng PY, Kumar A, Cavalli V, Klyachko VA. Circuit-based intervention corrects excessive dentate gyrus output in the fragile X mouse model. eLife 2024; 12:RP92563. [PMID: 38345852 PMCID: PMC10942577 DOI: 10.7554/elife.92563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Abnormal cellular and circuit excitability is believed to drive many core phenotypes in fragile X syndrome (FXS). The dentate gyrus is a brain area performing critical computations essential for learning and memory. However, little is known about dentate circuit defects and their mechanisms in FXS. Understanding dentate circuit dysfunction in FXS has been complicated by the presence of two types of excitatory neurons, the granule cells and mossy cells. Here we report that loss of FMRP markedly decreased excitability of dentate mossy cells, a change opposite to all other known excitability defects in excitatory neurons in FXS. This mossy cell hypo-excitability is caused by increased Kv7 function in Fmr1 knockout (KO) mice. By reducing the excitatory drive onto local hilar interneurons, hypo-excitability of mossy cells results in increased excitation/inhibition ratio in granule cells and thus paradoxically leads to excessive dentate output. Circuit-wide inhibition of Kv7 channels in Fmr1 KO mice increases inhibitory drive onto granule cells and normalizes the dentate output in response to physiologically relevant theta-gamma coupling stimulation. Our study suggests that circuit-based interventions may provide a promising strategy in this disorder to bypass irreconcilable excitability defects in different cell types and restore their pathophysiological consequences at the circuit level.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| | - Ajeet Kumar
- Department of Neuroscience, Washington University School of MedicineSt LouisUnited States
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of MedicineSt LouisUnited States
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
18
|
Soldovieri MV, Ambrosino P, Mosca I, Servettini I, Pietrunti F, Belperio G, Syrbe S, Taglialatela M, Lemke JR. De novo variants in KCNA3 cause developmental and epileptic encephalopathy. Ann Neurol 2024; 95:365-376. [PMID: 37964487 DOI: 10.1002/ana.26826] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 10/06/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVE Variants in several potassium channel genes, including KCNA1 and KCNA2, cause Developmental and Epileptic Encephalopathies (DEEs). We investigated whether variants in KCNA3, another mammalian homologue of the Drosophila shaker family and encoding for Kv1.3 subunits, can cause DEE. METHODS Genetic analysis of study individuals was performed by routine exome or genome sequencing, usually of parent-offspring trios. Phenotyping was performed via a standard clinical questionnaire. Currents from wild-type and/or mutant Kv1.3 subunits were investigated by whole-cell patch-clamp upon their heterologous expression. RESULTS Fourteen individuals, each carrying a de novo heterozygous missense variant in KCNA3, were identified. Most (12/14; 86%) had DEE with marked speech delay with or without motor delay, intellectual disability, epilepsy, and autism spectrum disorder. Functional analysis of Kv1.3 channels carrying each variant revealed heterogeneous functional changes, ranging from "pure" loss-of-function (LoF) effects due to faster inactivation kinetics, depolarized voltage-dependence of activation, slower activation kinetics, increased current inactivation, reduced or absent currents with or without dominant-negative effects, to "mixed" loss- and gain-of-function (GoF) effects. Compared to controls, Kv1.3 currents in lymphoblasts from 1 of the proband displayed functional changes similar to those observed upon heterologous expression of channels carrying the same variant. The antidepressant drug fluoxetine inhibited with similar potency the currents from wild-type and 1 of the Kv1.3 GoF variant. INTERPRETATION We describe a novel association of de novo missense variants in KCNA3 with a human DEE, and provide evidence that fluoxetine might represent a potential targeted treatment for individuals carrying variants with significant GoF effects. ANN NEUROL 2024;95:365-376.
Collapse
Affiliation(s)
| | - Paolo Ambrosino
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Ilaria Mosca
- Department of Medicine and Health Science "V. Tiberio", University of Molise, Campobasso, Italy
| | - Ilenio Servettini
- Department of Medicine and Health Science "V. Tiberio", University of Molise, Campobasso, Italy
| | - Francesca Pietrunti
- Department of Medicine and Health Science "V. Tiberio", University of Molise, Campobasso, Italy
| | - Giorgio Belperio
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Steffen Syrbe
- Center for Pediatrics and Adolescent Medicine, Division of Pediatric Epileptology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maurizio Taglialatela
- Department of Neuroscience, Division of Pharmacology, University of Naples "Federico II", Naples, Italy
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
19
|
Rai G, Sharma S, Bhasin J, Aggarwal K, Ahuja A, Dang S. Nanotechnological advances in the treatment of epilepsy: a comprehensive review. NANOTECHNOLOGY 2024; 35:152002. [PMID: 38194705 DOI: 10.1088/1361-6528/ad1c95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/09/2024] [Indexed: 01/11/2024]
Abstract
Epilepsy is one of the most prevalent chronic neurological disorders characterized by frequent unprovoked epileptic seizures. Epileptic seizures can develop from a broad range of underlying abnormalities such as tumours, strokes, infections, traumatic brain injury, developmental abnormalities, autoimmune diseases, and genetic predispositions. Sometimes epilepsy is not easily diagnosed and treated due to the large diversity of symptoms. Undiagnosed and untreated seizures deteriorate over time, impair cognition, lead to injuries, and can sometimes result in death. This review gives details about epilepsy, its classification on the basis of International League Against Epilepsy, current therapeutics which are presently offered for the treatment of epilepsy. Despite of the fact that more than 30 different anti-epileptic medication and antiseizure drugs are available, large number of epileptic patients fail to attain prolonged seizure independence. Poor onsite bioavailability of drugs due to blood brain barrier poses a major challenge in drug delivery to brain. The present review covers the limitations with the state-of-the-art strategies for managing seizures and emphasizes the role of nanotechnology in overcoming these issues. Various nano-carriers like polymeric nanoparticles, dendrimers, lipidic nanoparticles such as solid lipid nanoparticles, nano-lipid carriers, have been explored for the delivery of anti-epileptic drugs to brain using oral and intranasal routes. Nano-carries protect the encapsulated drugs from degradation and provide a platform to deliver controlled release over prolonged periods, improved permeability and bioavailability at the site of action. The review also emphasises in details about the role of neuropeptides for the treatment of epilepsy.
Collapse
Affiliation(s)
- Garima Rai
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Surbhi Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Jasveen Bhasin
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Kanica Aggarwal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Alka Ahuja
- College of Pharmacy, National University of Science and Technology, Muscat, Oman
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| |
Collapse
|
20
|
Zhang R, Xie J, Yuan X, Yu Y, Zhuang Y, Zhang F, Hou J, Liu Y, Huang W, Zhang M, Li J, Gong Q, Peng X. Newly discovered variants in unexplained neonatal encephalopathy. Mol Genet Genomic Med 2024; 12:e2354. [PMID: 38284441 PMCID: PMC10795097 DOI: 10.1002/mgg3.2354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND The genetic background of neonatal encephalopathy (NE) is complicated and early diagnosis is beneficial to optimizing therapeutic strategy for patients. METHODS NE Patients with unclear etiology received regular clinical tests including ammonia test, metabolic screening test, amplitude-integrated electroencephalographic (aEEG) monitoring, brain Magnetic Resonance Imaging (MRI) scanning, and genetic test. The protein structure change was predicted using Dynamut2 and RoseTTAFold. RESULTS 15 out of a total of 113 NE Patients were detected with newly reported pathogenic variants. In this sub-cohort, (1) seizure was the primary initial symptoms; (2) four patients had abnormal metabolic screening results, and two of them were also diagnosed with excessive blood ammonia concentration; (3) the brain MRI results were irregular in three infants and the brain waves were of moderate-severe abnormality in about a half of the patients. The novel pathogenic variants discovered in this study belonged to 12 genes, and seven of them were predicted to introduce a premature translation termination. In-silicon predictions showed that four variants were destructive to the protein structure of KCNQ2. CONCLUSION Our study expands the mutation spectrum of genes associated with NE and introduces new evidence for molecular diagnosis in this newborn illness.
Collapse
Affiliation(s)
- Rong Zhang
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Jingjing Xie
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Xiao Yuan
- Department of Laboratory DiagnosisChangsha Kingmed Center for Clinical LaboratoryChangshaHunanChina
| | - Yan Yu
- Department of Laboratory DiagnosisChangsha Kingmed Center for Clinical LaboratoryChangshaHunanChina
| | - Yan Zhuang
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Fan Zhang
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Jianfei Hou
- Department of Laboratory DiagnosisChangsha Kingmed Center for Clinical LaboratoryChangshaHunanChina
| | - Yanqin Liu
- Department of Laboratory DiagnosisChangsha Kingmed Center for Clinical LaboratoryChangshaHunanChina
| | - Weiqing Huang
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Min Zhang
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Junshuai Li
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| | - Qiang Gong
- Department of Laboratory DiagnosisChangsha Kingmed Center for Clinical LaboratoryChangshaHunanChina
| | - Xiaoming Peng
- Department of NeonatologyHunan Children's HospitalChangshaHunanChina
| |
Collapse
|
21
|
Mínguez-Viñas T, Prakash V, Wang K, Lindström SH, Pozzi S, Scott SA, Spiteri E, Stevenson DA, Ashley EA, Gunnarsson C, Pantazis A. Two epilepsy-associated variants in KCNA2 (K V 1.2) at position H310 oppositely affect channel functional expression. J Physiol 2023; 601:5367-5389. [PMID: 37883018 DOI: 10.1113/jp285052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
Two KCNA2 variants (p.H310Y and p.H310R) were discovered in paediatric patients with epilepsy and developmental delay. KCNA2 encodes KV 1.2-channel subunits, which regulate neuronal excitability. Both gain and loss of KV 1.2 function cause epilepsy, precluding the prediction of variant effects; and while H310 is conserved throughout the KV -channel superfamily, it is largely understudied. We investigated both variants in heterologously expressed, human KV 1.2 channels by immunocytochemistry, electrophysiology and voltage-clamp fluorometry. Despite affecting the same channel, at the same position, and being associated with severe neurological disease, the two variants had diametrically opposite effects on KV 1.2 functional expression. The p.H310Y variant produced 'dual gain of function', increasing both cell-surface trafficking and activity, delaying channel closure. We found that the latter is due to the formation of a hydrogen bond that stabilizes the active state of the voltage-sensor domain. Additionally, H310Y abolished 'ball and chain' inactivation of KV 1.2 by KV β1 subunits, enhancing gain of function. In contrast, p.H310R caused 'dual loss of function', diminishing surface levels by multiple impediments to trafficking and inhibiting voltage-dependent channel opening. We discuss the implications for KV -channel biogenesis and function, an emergent hotspot for disease-associated variants, and mechanisms of epileptogenesis. KEY POINTS: KCNA2 encodes the subunits of KV 1.2 voltage-activated, K+ -selective ion channels, which regulate electrical signalling in neurons. We characterize two KCNA2 variants from patients with developmental delay and epilepsy. Both variants affect position H310, highly conserved in KV channels. The p.H310Y variant caused 'dual gain of function', increasing both KV 1.2-channel activity and the number of KV 1.2 subunits on the cell surface. H310Y abolished 'ball and chain' (N-type) inactivation of KV 1.2 by KV β1 subunits, enhancing the gain-of-function phenotype. The p.H310R variant caused 'dual loss of function', diminishing the presence of KV 1.2 subunits on the cell surface and inhibiting voltage-dependent channel opening. As H310Y stabilizes the voltage-sensor active conformation and abolishes N-type inactivation, it can serve as an investigative tool for functional and pharmacological studies.
Collapse
Affiliation(s)
- Teresa Mínguez-Viñas
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Varsha Prakash
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kaiqian Wang
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sarah H Lindström
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Serena Pozzi
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Stuart A Scott
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Elizabeth Spiteri
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - David A Stevenson
- Division of Medical Genetics, Stanford University, Palo Alto, California, USA
| | - Euan A Ashley
- Division of Medical Genetics, Stanford University, Palo Alto, California, USA
| | - Cecilia Gunnarsson
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Genetics, Linköping University, Linköping, Sweden
- Centre for Rare Diseases in South East Region of Sweden, Linköping University, Linköping, Sweden
| | - Antonios Pantazis
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
Shorthouse D, Zhuang L, Rahrmann EP, Kosmidou C, Wickham Rahrmann K, Hall M, Greenwood BM, Devonshire G, Gilbertson RJ, Fitzgerald RC, Hall BA. KCNQ potassium channels modulate Wnt activity in gastro-oesophageal adenocarcinomas. Life Sci Alliance 2023; 6:e202302124. [PMID: 37748809 PMCID: PMC10520261 DOI: 10.26508/lsa.202302124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Voltage-sensitive potassium channels play an important role in controlling membrane potential and ionic homeostasis in the gut and have been implicated in gastrointestinal (GI) cancers. Through large-scale analysis of 897 patients with gastro-oesophageal adenocarcinomas (GOAs) coupled with in vitro models, we find KCNQ family genes are mutated in ∼30% of patients, and play therapeutically targetable roles in GOA cancer growth. KCNQ1 and KCNQ3 mediate the WNT pathway and MYC to increase proliferation through resultant effects on cadherin junctions. This also highlights novel roles of KCNQ3 in non-excitable tissues. We also discover that activity of KCNQ3 sensitises cancer cells to existing potassium channel inhibitors and that inhibition of KCNQ activity reduces proliferation of GOA cancer cells. These findings reveal a novel and exploitable role of potassium channels in the advancement of human cancer, and highlight that supplemental treatments for GOAs may exist through KCNQ inhibitors.
Collapse
Affiliation(s)
- David Shorthouse
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| | - Lizhe Zhuang
- Institute for Early Detection, CRUK Cambridge Centre, Cambridge, UK
| | - Eric P Rahrmann
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | | | - Michael Hall
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Benedict M Greenwood
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| | - Ginny Devonshire
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Richard J Gilbertson
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, London, UK
| |
Collapse
|
23
|
Deng PY, Kumar A, Cavalli V, Klyachko VA. Circuit-based intervention corrects excessive dentate gyrus output in the Fragile X mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559792. [PMID: 37808793 PMCID: PMC10557679 DOI: 10.1101/2023.09.27.559792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Abnormal cellular and circuit excitability is believed to drive many core phenotypes in fragile X syndrome (FXS). The dentate gyrus is a brain area performing critical computations essential for learning and memory. However, little is known about dentate circuit defects and their mechanisms in FXS. Understanding dentate circuit dysfunction in FXS has been complicated by the presence of two types of excitatory neurons, the granule cells and mossy cells. Here we report that loss of FMRP markedly decreased excitability of dentate mossy cells, a change opposite to all other known excitability defects in excitatory neurons in FXS. This mossy cell hypo-excitability is caused by increased Kv7 function in Fmr1 KO mice. By reducing the excitatory drive onto local hilar interneurons, hypo-excitability of mossy cells results in increased excitation/inhibition ratio in granule cells and thus paradoxically leads to excessive dentate output. Circuit-wide inhibition of Kv7 channels in Fmr1 KO mice increases inhibitory drive onto granule cells and normalizes the dentate output in response to physiologically relevant theta-gamma coupling stimulation. Our study suggests that circuit-based interventions may provide a promising strategy in this disorder to bypass irreconcilable excitability defects in different cell types and restore their pathophysiological consequences at the circuit level.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Ajeet Kumar
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Vitaly A. Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| |
Collapse
|
24
|
Bortolami A, Sesti F. Ion channels in neurodevelopment: lessons from the Integrin-KCNB1 channel complex. Neural Regen Res 2023; 18:2365-2369. [PMID: 37282454 PMCID: PMC10360111 DOI: 10.4103/1673-5374.371347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Ion channels modulate cellular excitability by regulating ionic fluxes across biological membranes. Pathogenic mutations in ion channel genes give rise to epileptic disorders that are among the most frequent neurological diseases affecting millions of individuals worldwide. Epilepsies are triggered by an imbalance between excitatory and inhibitory conductances. However, pathogenic mutations in the same allele can give rise to loss-of-function and/or gain-of-function variants, all able to trigger epilepsy. Furthermore, certain alleles are associated with brain malformations even in the absence of a clear electrical phenotype. This body of evidence argues that the underlying epileptogenic mechanisms of ion channels are more diverse than originally thought. Studies focusing on ion channels in prenatal cortical development have shed light on this apparent paradox. The picture that emerges is that ion channels play crucial roles in landmark neurodevelopmental processes, including neuronal migration, neurite outgrowth, and synapse formation. Thus, pathogenic channel mutants can not only cause epileptic disorders by altering excitability, but further, by inducing morphological and synaptic abnormalities that are initiated during neocortex formation and may persist into the adult brain.
Collapse
Affiliation(s)
- Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, West Piscataway, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, West Piscataway, NJ, USA
| |
Collapse
|
25
|
Zheng JY, Kang T, Jiang C, Lin LK, Gao L, Jin LH, Shu Y, Zhang JJ, Li C, Chen B, Shen YH. Gut microbiome and brain transcriptome analyses reveal the effect of walnut oil in preventing scopolamine-induced cognitive impairment. Food Funct 2023; 14:9707-9724. [PMID: 37814808 DOI: 10.1039/d3fo01893h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Walnut Oil (WO) is recognized for its potential to improve cognition, but the mechanisms of its action related to improving cognitive impairment are not yet clear. In this study, the components of walnut oil were measured, and it was found that WO supplementation for 8 weeks could significantly prevent cognitive behavioral deficits and synaptic dysfunction induced by intraperitoneal injection of scopolamine (SCOP) in mice. By comparing and analyzing the changes in the hippocampal synaptic structure, oxidative stress, neurotransmitter fluctuations, brain transcriptome, inflammatory factors and gut microbiota in mice from different treatment groups, we observed a significant correlation between synaptic transmission genes, gut microbiota and neurotransmission in the WO supplemented group. It was found that WO supplementation could influence the secretion of neurotransmitters Ach and 5-HT by modulating the gut microbiota in vivo, thereby improving cognitive impairment through the central nervous system and hypothalamic-pituitary-adrenal (HPA) axis regulation.
Collapse
Affiliation(s)
- Jing Yi Zheng
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Ting Kang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Chao Jiang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Li Ke Lin
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Lu Gao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Li Hua Jin
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Yu Shu
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China
| | - Jing Jing Zhang
- School of Chemical Engineering, Northwest University, Xi'an, Shaanxi 710069, China
| | - Cong Li
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Bang Chen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| | - Ye Hua Shen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, National Demonstration Center for Experimental Chemistry Education, Northwest University, Xi'an, Shaanxi 710127, China.
| |
Collapse
|
26
|
Liu YC, So EC, Wu SN. Cannabidiol Modulates M-Type K + and Hyperpolarization-Activated Cation Currents. Biomedicines 2023; 11:2651. [PMID: 37893024 PMCID: PMC10604323 DOI: 10.3390/biomedicines11102651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Cannabidiol (CBD) is a naturally occurring compound found in the Cannabis plant that is known for its potential therapeutic effects. However, its impact on membrane ionic currents remains a topic of debate. This study aimed to investigate how CBD modifies various types of ionic currents in pituitary GH3 cells. Results showed that exposure to CBD led to a concentration-dependent decrease in M-type K+ currents (IK(M)), with an IC50 of 3.6 μM, and caused the quasi-steady-state activation curve of the current to shift to a more depolarized potential with no changes in the curve's steepness. The CBD-mediated block of IK(M) was not reversed by naloxone, suggesting that it was not mediated by opioid receptors. The IK(M) elicited by pulse-train stimulation was also decreased upon exposure to CBD. The magnitude of erg-mediated K+ currents was slightly reduced by adding CBD (10 μM), while the density of voltage-gated Na+ currents elicited by a short depolarizing pulse was not affected by it. Additionally, CBD decreased the magnitude of hyperpolarization-activated cation currents (Ih) with an IC50 of 3.3 μM, and the decrease was reversed by oxaliplatin. The quasi-steady-state activation curve of Ih was shifted in the leftward direction with no changes in the slope factor of the curve. CBD also diminished the strength of voltage-dependent hysteresis on Ih elicited by upright isosceles-triangular ramp voltage. Collectively, these findings suggest that CBD's modification of ionic currents presented herein is independent of cannabinoid or opioid receptors and may exert a significant impact on the functional activities of excitable cells occurring in vitro or in vivo.
Collapse
Affiliation(s)
- Yen-Chin Liu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Department of Anesthesiology, School of Post-Baccalaureate, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Edmund Cheung So
- Department of Anesthesia, An-Nan Hospital, China Medical University, Tainan 70965, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- School of Medicine, National Sun-Yat Sen University College of Medicine, Kaohsiung 80424, Taiwan
- Department of Research and Education, An-Nan Hospital, China Medical University, Tainan 70965, Taiwan
| |
Collapse
|
27
|
Varghese N, Moscoso B, Chavez A, Springer K, Ortiz E, Soh H, Santaniello S, Maheshwari A, Tzingounis AV. KCNQ2/3 Gain-of-Function Variants and Cell Excitability: Differential Effects in CA1 versus L2/3 Pyramidal Neurons. J Neurosci 2023; 43:6479-6494. [PMID: 37607817 PMCID: PMC10513074 DOI: 10.1523/jneurosci.0980-23.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023] Open
Abstract
Gain-of-function (GOF) pathogenic variants in the potassium channels KCNQ2 and KCNQ3 lead to hyperexcitability disorders such as epilepsy and autism spectrum disorders. However, the underlying cellular mechanisms of how these variants impair forebrain function are unclear. Here, we show that the R201C variant in KCNQ2 has opposite effects on the excitability of two types of mouse pyramidal neurons of either sex, causing hyperexcitability in layer 2/3 (L2/3) pyramidal neurons and hypoexcitability in CA1 pyramidal neurons. Similarly, the homologous R231C variant in KCNQ3 leads to hyperexcitability in L2/3 pyramidal neurons and hypoexcitability in CA1 pyramidal neurons. However, the effects of KCNQ3 gain-of-function on excitability are specific to superficial CA1 pyramidal neurons. These findings reveal a new level of complexity in the function of KCNQ2 and KCNQ3 channels in the forebrain and provide a framework for understanding the effects of gain-of-function variants and potassium channels in the brain.SIGNIFICANCE STATEMENT KCNQ2/3 gain-of-function (GOF) variants lead to severe forms of neurodevelopmental disorders, but the mechanisms by which these channels affect neuronal activity are poorly understood. In this study, using a series of transgenic mice we demonstrate that the same KCNQ2/3 GOF variants can lead to either hyperexcitability or hypoexcitability in different types of pyramidal neurons [CA1 vs layer (L)2/3]. Additionally, we show that expression of the recurrent KCNQ2 GOF variant R201C in forebrain pyramidal neurons could lead to seizures and SUDEP. Our data suggest that the effects of KCNQ2/3 GOF variants depend on specific cell types and brain regions, possibly accounting for the diverse range of phenotypes observed in individuals with KCNQ2/3 GOF variants.
Collapse
Affiliation(s)
- Nissi Varghese
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269
| | - Bruno Moscoso
- Department of Neurology, Baylor College of Medicine, Houston, Texas 77030
| | - Ana Chavez
- Department of Neurology, Baylor College of Medicine, Houston, Texas 77030
| | - Kristen Springer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269
| | - Erika Ortiz
- Department of Neurology, Baylor College of Medicine, Houston, Texas 77030
| | - Heun Soh
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269
| | - Sabato Santaniello
- Department of Biomedical Engineering and Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Atul Maheshwari
- Department of Neurology, Baylor College of Medicine, Houston, Texas 77030
| | - Anastasios V Tzingounis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269
| |
Collapse
|
28
|
Yang GM, Tian FY, Shen YW, Yang CY, Yuan H, Li P, Gao ZB. Functional characterization and in vitro pharmacological rescue of KCNQ2 pore mutations associated with epileptic encephalopathy. Acta Pharmacol Sin 2023; 44:1589-1599. [PMID: 36932231 PMCID: PMC10374643 DOI: 10.1038/s41401-023-01073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/26/2023] [Indexed: 03/19/2023]
Abstract
Mutations in the KCNQ2 gene encoding KV7.2 subunit that mediates neuronal M-current cause a severe form of developmental and epileptic encephalopathy (DEE). Electrophysiological evaluation of KCNQ2 mutations has been proved clinically useful in improving outcome prediction and choosing rational anti-seizure medications (ASMs). In this study we described the clinical characteristics, electrophysiological phenotypes and the in vitro response to KCNQ openers of five KCNQ2 pore mutations (V250A, N258Y, H260P, A265T and G290S) from seven patients diagnosed with KCNQ2-DEE. The KCNQ2 variants were transfected into Chinese hamster ovary (CHO) cells alone, in combination with KCNQ3 (1:1) or with wild-type KCNQ2 (KCNQ2-WT) and KCNQ3 in a ratio of 1:1:2, respectively. Their expression and electrophysiological function were assessed. When transfected alone or in combination with KCNQ3, none of these mutations affected the membrane expression of KCNQ2, but most failed to induce a potassium current except A265T, in which trace currents were observed when co-transfected with KCNQ3. When co-expressed with KCNQ2-WT and KCNQ3 (1:1:2), the currents at 0 mV of these mutations were decreased by 30%-70% compared to the KCNQ2/3 channel, which could be significantly rescued by applying KCNQ openers including the approved antiepileptic drug retigabine (RTG, 10 μM), as well as two candidates subjected to clinical trials, pynegabine (HN37, 1 μM) and XEN1101 (1 μM). These newly identified pathologic variants enrich the KCNQ2-DEE mutation hotspots in the pore-forming domain. This electrophysiological study provides a rational basis for personalized therapy with KCNQ openers in DEE patients carrying loss-of-function (LOF) mutations in KCNQ2.
Collapse
Affiliation(s)
- Gui-Mei Yang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Fu-Yun Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Yan-Wen Shen
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Beijing, 100853, China
- Department of Pediatric neurology, Children's Hospital of Fudan university at Xiamen, Xiamen, 361006, China
| | - Chuan-Yan Yang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hui Yuan
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Ping Li
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Zhao-Bing Gao
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
29
|
Cioclu MC, Mosca I, Ambrosino P, Puzo D, Bayat A, Wortmann SB, Koch J, Strehlow V, Shirai K, Matsumoto N, Sanders SJ, Michaud V, Legendre M, Riva A, Striano P, Muhle H, Pendziwiat M, Lesca G, Mangano GD, Nardello R, Lemke JR, Møller RS, Soldovieri MV, Rubboli G, Taglialatela M. KCNT2-Related Disorders: Phenotypes, Functional, and Pharmacological Properties. Ann Neurol 2023; 94:332-349. [PMID: 37062836 DOI: 10.1002/ana.26662] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
OBJECTIVE Pathogenic variants in KCNT2 are rare causes of developmental epileptic encephalopathy (DEE). We herein describe the phenotypic and genetic features of patients with KCNT2-related DEE, and the in vitro functional and pharmacological properties of KCNT2 channels carrying 14 novel or previously untested variants. METHODS Twenty-five patients harboring KCNT2 variants were investigated: 12 were identified through an international collaborative network, 13 were retrieved from the literature. Clinical data were collected and included in a standardized phenotyping sheet. Novel variants were detected using exome sequencing and classified using ACMG criteria. Functional and pharmacological studies were performed by whole-cell electrophysiology in HEK-293 and SH-SY5Y cells. RESULTS The phenotypic spectrum encompassed: (a) intellectual disability/developmental delay (21/22 individuals with available information), ranging from mild to severe/profound; (b) epilepsy (15/25); (c) neurological impairment, with altered muscle tone (14/22); (d) dysmorphisms (13/20). Nineteen pathogenic KCNT2 variants were found (9 new, 10 reported previously): 16 missense, 1 in-frame deletion of a single amino acid, 1 nonsense, and 1 frameshift. Among tested variants, 8 showed gain-of-function (GoF), and 6 loss-of-function (LoF) features when expressed heterologously in vitro. Quinidine and fluoxetine blocked all GoF variants, whereas loxapine and riluzole activated some LoF variants while blocking others. INTERPRETATION We expanded the phenotypic and genotypic spectrum of KCNT2-related disorders, highlighting novel genotype-phenotype associations. Pathogenic KCNT2 variants cause GoF or LoF in vitro phenotypes, and each shows a unique pharmacological profile, suggesting the need for in vitro functional and pharmacological investigation to enable targeted therapies based on the molecular phenotype. ANN NEUROL 2023;94:332-349.
Collapse
Affiliation(s)
- Maria Cristina Cioclu
- Department of Epilepsy Genetics and Personalized Medicine (member of ERN EpiCARE), Danish Epilepsy Centre, Dianalund, Denmark
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Ilaria Mosca
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Paolo Ambrosino
- Dept. of Science and Technology, University of Sannio, Benevento, Italy
| | - Deborah Puzo
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Allan Bayat
- Department of Epilepsy Genetics and Personalized Medicine (member of ERN EpiCARE), Danish Epilepsy Centre, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Saskia B Wortmann
- University Children's Hospital, Paracelsus Medical University, Salzburg, Austria
- Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Johannes Koch
- University Children's Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Vincent Strehlow
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Kentaro Shirai
- Department of Pediatrics, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Vincent Michaud
- Service de Génétique Médicale, Centre de Référence Anomalies du Développement et Syndrome Malformatifs, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- Maladies rares: Génétique et Métabolisme (MRGM), INSERM U1211, Université de Bordeaux, Bordeaux, France
| | - Marine Legendre
- Service de Génétique Médicale, Centre de Référence Anomalies du Développement et Syndrome Malformatifs, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Antonella Riva
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Hiltrud Muhle
- Department of Neuropediatrics, University Medical Centre Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Manuela Pendziwiat
- Department of Neuropediatrics, University Medical Centre Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Gaetan Lesca
- Pathophysiology and Genetics of Neuron and Muscle (PNMG), UCBL, CNRS UMR5261-INSERM U1315, Lyon, France
- Department of Medical Genetics, University Hospital of Lyon and Claude Bernard Lyon I University, Lyon, France
| | - Giuseppe Donato Mangano
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Rosaria Nardello
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine (member of ERN EpiCARE), Danish Epilepsy Centre, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Maria Virginia Soldovieri
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Guido Rubboli
- Department of Epilepsy Genetics and Personalized Medicine (member of ERN EpiCARE), Danish Epilepsy Centre, Dianalund, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
30
|
Hou B, Santaniello S, Tzingounis AV. KCNQ2 channels regulate the population activity of neonatal GABAergic neurons ex vivo. Front Neurol 2023; 14:1207539. [PMID: 37409016 PMCID: PMC10318362 DOI: 10.3389/fneur.2023.1207539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/18/2023] [Indexed: 07/07/2023] Open
Abstract
Over the last decade KCNQ2 channels have arisen as fundamental and indispensable regulators of neonatal brain excitability, with KCNQ2 loss-of-function pathogenic variants being increasingly identified in patients with developmental and epileptic encephalopathy. However, the mechanisms by which KCNQ2 loss-of-function variants lead to network dysfunction are not fully known. An important remaining knowledge gap is whether loss of KCNQ2 function alters GABAergic interneuron activity early in development. To address this question, we applied mesoscale calcium imaging ex vivo in postnatal day 4-7 mice lacking KCNQ2 channels in interneurons (Vgat-ires-cre;Kcnq2f/f;GCamp5). In the presence of elevated extracellular potassium concentrations, ablation of KCNQ2 channels from GABAergic cells increased the interneuron population activity in the hippocampal formation and regions of the neocortex. We found that this increased population activity depends on fast synaptic transmission, with excitatory transmission promoting the activity and GABAergic transmission curtailing it. Together, our data show that loss of function of KCNQ2 channels from interneurons increases the network excitability of the immature GABAergic circuits, revealing a new function of KCNQ2 channels in interneuron physiology in the developing brain.
Collapse
Affiliation(s)
- Bowen Hou
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Sabato Santaniello
- Department of Biomedical Engineering and CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
| | - Anastasios V. Tzingounis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Department of Biomedical Engineering and CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
31
|
Oh H, Lee S, Oh Y, Kim S, Kim YS, Yang Y, Choi W, Yoo YE, Cho H, Lee S, Yang E, Koh W, Won W, Kim R, Lee CJ, Kim H, Kang H, Kim JY, Ku T, Paik SB, Kim E. Kv7/KCNQ potassium channels in cortical hyperexcitability and juvenile seizure-related death in Ank2-mutant mice. Nat Commun 2023; 14:3547. [PMID: 37321992 PMCID: PMC10272139 DOI: 10.1038/s41467-023-39203-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
Autism spectrum disorders (ASD) represent neurodevelopmental disorders characterized by social deficits, repetitive behaviors, and various comorbidities, including epilepsy. ANK2, which encodes a neuronal scaffolding protein, is frequently mutated in ASD, but its in vivo functions and disease-related mechanisms are largely unknown. Here, we report that mice with Ank2 knockout restricted to cortical and hippocampal excitatory neurons (Ank2-cKO mice) show ASD-related behavioral abnormalities and juvenile seizure-related death. Ank2-cKO cortical neurons show abnormally increased excitability and firing rate. These changes accompanied decreases in the total level and function of the Kv7.2/KCNQ2 and Kv7.3/KCNQ3 potassium channels and the density of these channels in the enlengthened axon initial segment. Importantly, the Kv7 agonist, retigabine, rescued neuronal excitability, juvenile seizure-related death, and hyperactivity in Ank2-cKO mice. These results suggest that Ank2 regulates neuronal excitability by regulating the length of and Kv7 density in the AIS and that Kv7 channelopathy is involved in Ank2-related brain dysfunctions.
Collapse
Affiliation(s)
- Hyoseon Oh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yusang Oh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Korea
| | - Seongbin Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Young Seo Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
| | - Yeji Yang
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanjiro, Ochang, Cheongju, Chungbuk, 28119, Korea
| | - Woochul Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Korea
| | - Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Heejin Cho
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Seungjoon Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Esther Yang
- Department of Anatomy and Brain Korea 21 Graduate Program, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, IBS, Daejeon, 34126, Korea
| | - Woojin Won
- Center for Cognition and Sociality, IBS, Daejeon, 34126, Korea
| | - Ryunhee Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, IBS, Daejeon, 34126, Korea
| | - Hyun Kim
- Department of Anatomy and Brain Korea 21 Graduate Program, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanjiro, Ochang, Cheongju, Chungbuk, 28119, Korea
| | - Taeyun Ku
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
| |
Collapse
|
32
|
Garrido JJ. Contribution of Axon Initial Segment Structure and Channels to Brain Pathology. Cells 2023; 12:cells12081210. [PMID: 37190119 DOI: 10.3390/cells12081210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Brain channelopathies are a group of neurological disorders that result from genetic mutations affecting ion channels in the brain. Ion channels are specialized proteins that play a crucial role in the electrical activity of nerve cells by controlling the flow of ions such as sodium, potassium, and calcium. When these channels are not functioning properly, they can cause a wide range of neurological symptoms such as seizures, movement disorders, and cognitive impairment. In this context, the axon initial segment (AIS) is the site of action potential initiation in most neurons. This region is characterized by a high density of voltage-gated sodium channels (VGSCs), which are responsible for the rapid depolarization that occurs when the neuron is stimulated. The AIS is also enriched in other ion channels, such as potassium channels, that play a role in shaping the action potential waveform and determining the firing frequency of the neuron. In addition to ion channels, the AIS contains a complex cytoskeletal structure that helps to anchor the channels in place and regulate their function. Therefore, alterations in this complex structure of ion channels, scaffold proteins, and specialized cytoskeleton may also cause brain channelopathies not necessarily associated with ion channel mutations. This review will focus on how the AISs structure, plasticity, and composition alterations may generate changes in action potentials and neuronal dysfunction leading to brain diseases. AIS function alterations may be the consequence of voltage-gated ion channel mutations, but also may be due to ligand-activated channels and receptors and AIS structural and membrane proteins that support the function of voltage-gated ion channels.
Collapse
Affiliation(s)
- Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28002 Madrid, Spain
| |
Collapse
|
33
|
Okamura Y, Yoshioka D. What voltage-sensing phosphatases can reveal about the mechanisms of ion channel regulation by phosphoinositides. Biochem Soc Trans 2023; 51:827-839. [PMID: 37052219 DOI: 10.1042/bst20221065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023]
Abstract
Many membrane proteins including ion channels and ion transporters are regulated by membrane phospholipids such as phosphoinositides in cell membranes and organelles. Voltage-sensing phosphatase, VSP, is a voltage-sensitive phosphoinositide phosphatase which dephosphorylates PI(4,5)P2 into PI(4)P. VSP rapidly reduces the level of PI(4,5)P2 upon membrane depolarization, thus serving as a useful tool to quantitatively study phosphoinositide-regulation of ion channels and ion transporters using a cellular electrophysiology system. In this review, we focus on the application of VSPs to Kv7 family potassium channels, which have been important research targets in biophysics, pharmacology and medicine.
Collapse
Affiliation(s)
- Yasushi Okamura
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Yamada Oka 2-2, Suita, Osaka 565-0871, Japan
| | - Daisuke Yoshioka
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Yamada Oka 2-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
34
|
Pruunsild P, Bengtson CP, Loss I, Lohrer B, Bading H. Expression of the primate-specific LINC00473 RNA in mouse neurons promotes excitability and CREB-regulated transcription. J Biol Chem 2023; 299:104671. [PMID: 37019214 DOI: 10.1016/j.jbc.2023.104671] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
The LINC00473 (Lnc473) gene has previously been shown to be associated with cancer and psychiatric disorders. Its expression is elevated in several types of tumors and decreased in the brains of patients diagnosed with schizophrenia or major depression. In neurons, Lnc473 transcription is strongly responsive to synaptic activity, suggesting a role in adaptive, plasticity-related mechanisms. However, the function of Lnc473 is largely unknown. Here, using a recombinant adeno-associated viral vector, we introduced a primate-specific human Lnc473 RNA into mouse primary neurons. We show that this resulted in a transcriptomic shift comprising downregulation of epilepsy-associated genes and a rise in cAMP response element binding protein (CREB) activity, which was driven by augmented CREB-regulated transcription coactivator 1 (CRTC1) nuclear localization. Moreover, we demonstrate that ectopic Lnc473 expression increased neuronal excitability as well as network excitability. These findings suggest that primates may possess a lineage-specific activity-dependent modulator of CREB-regulated neuronal excitability.
Collapse
|
35
|
Cossu A, Lo Barco T, Proietti J, Dalla Bernardina B, Cantalupo G, Ghobert L, Brambilla I, Giarola E, Costa A, De Benito T, Bethge S, Cardot S, Montwill I, Remonato E, Gramaglia S, Darra F. Clinical characteristics of 80 subjects with KCNQ2-related encephalopathy: Results from a family-driven survey. Epilepsy Behav 2023; 142:109153. [PMID: 36989566 DOI: 10.1016/j.yebeh.2023.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/31/2023]
Abstract
Variants of KCNQ2 are associated with a wide spectrum of disorders, ranging from Self-limiting Neonatal Epilepsy (SelNE) to Early Onset Developmental and Epileptic Encephalopathy (KCNQ2-DEE). Comorbidities associated with this end of the spectrum have been seldomly described and their impact on the life of patients and their families is yet to be investigated. Collaborating with caregivers from different European family associations, we have developed a questionnaire aimed at investigating the onset and frequency of epileptic seizures, anti-seizure medications (ASM), hospitalizations, stages of development, and comorbidities. Responses from 80 patients, 40 males, from 14 countries have been collected. Median age 7.6 years (4 months - 43.6 years). Of 76 epileptic patients (93.6%), 55.3% were seizure-free with a mean age at last seizure of 26.7 months. Among patients with active epilepsy, those older have a lower frequency of seizures (p > 0.05). We were able to identify three different clusters of varying severity (Mild, Severe, Profound), based on neurodevelopmental features and symptoms, excluding epilepsy. Patients in a higher severity cluster had a higher mean number of comorbidities, which had a higher impact on families. Notably, patients in different clusters presented different epilepsy onset and courses. This study constitutes the most extensive data collection of patients with KCNQ2-DEE, with a focus on comorbidities in a wide age group. The participation of caregivers helps to define the impact of the disease on the lives of patients and families and can help identify new primary and secondary outcomes beyond seizures in future studies.
Collapse
Affiliation(s)
- A Cossu
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - T Lo Barco
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - J Proietti
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - B Dalla Bernardina
- Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - G Cantalupo
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - L Ghobert
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy
| | - I Brambilla
- Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Alleanza Epilessie Rare e Complesse, Italy; Dravet Italia Onlus, Italy; European KCNQ2 Association Odv, Italy
| | - E Giarola
- Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Alleanza Epilessie Rare e Complesse, Italy; Dravet Italia Onlus, Italy
| | - A Costa
- European KCNQ2 Association Odv, Italy
| | | | | | - S Cardot
- KCNQ2 France Développement, France
| | | | - E Remonato
- Alleanza Epilessie Rare e Complesse, Italy; European KCNQ2 Association Odv, Italy
| | | | - F Darra
- Child Neuropsychiatry Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy; Research Center for Pediatric Epilepsies (CREP) - Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Italy.
| |
Collapse
|
36
|
Zahra A, Liu R, Han W, Meng H, Wang Q, Wang Y, Campbell SL, Wu J. K Ca-Related Neurological Disorders: Phenotypic Spectrum and Therapeutic Indications. Curr Neuropharmacol 2023; 21:1504-1518. [PMID: 36503451 PMCID: PMC10472807 DOI: 10.2174/1570159x21666221208091805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Although potassium channelopathies have been linked to a wide range of neurological conditions, the underlying pathogenic mechanism is not always clear, and a systematic summary of clinical manifestation is absent. Several neurological disorders have been associated with alterations of calcium-activated potassium channels (KCa channels), such as loss- or gain-of-function mutations, post-transcriptional modification, etc. Here, we outlined the current understanding of the molecular and cellular properties of three subtypes of KCa channels, including big conductance KCa channels (BK), small conductance KCa channels (SK), and the intermediate conductance KCa channels (IK). Next, we comprehensively reviewed the loss- or gain-of-function mutations of each KCa channel and described the corresponding mutation sites in specific diseases to broaden the phenotypic-genotypic spectrum of KCa-related neurological disorders. Moreover, we reviewed the current pharmaceutical strategies targeting KCa channels in KCa-related neurological disorders to provide new directions for drug discovery in anti-seizure medication.
Collapse
Affiliation(s)
- Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Wenzhe Han
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Hui Meng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - YunFu Wang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Susan L. Campbell
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
37
|
Zhan X, Drummond-Main C, Greening D, Yao J, Chen SWR, Appendino JP, Au PYB, Turner RW. Cannabidiol counters the effects of a dominant-negative pathogenic Kv7.2 variant. iScience 2022; 25:105092. [PMID: 36157585 PMCID: PMC9490039 DOI: 10.1016/j.isci.2022.105092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022] Open
Abstract
Epilepsy and neurodevelopmental disorders can arise from pathogenic variants of KCNQ (Kv7) channels. A patient with developmental and epileptic encephalopathy exhibited an in-frame deletion of histidine 260 on Kv7.2. Coexpression of Kv7.2 mutant (mut) subunits with Kv7.3 invoked a decrease in current density, a depolarizing shift in voltage for activation, and a decrease in membrane conductance. Biotinylation revealed an increased level of surface Kv7.2mut compared to Kv7.3 with no change in total membrane protein expression. Super-resolution and FRET imaging confirmed heteromeric channel formation and a higher expression density of Kv7.2mut. Cannabidiol (1 μM) offset the effects of Kv7.2mut by inducing a hyperpolarizing shift in voltage for activation independent of CB1 or CB2 receptors. These data reveal that the ability for cannabidiol to reduce the effects of a pathogenic Kv7.2 variant supports its use as a potential therapeutic to reduce seizure activity. A patient with epileptic encephalopathy exhibits a Kv7.2 deletion at H260 (Kv7.2mut) Kv7.2mut shows increased expression at the membrane compared to Kv7.3 Kv7.2mut acts in a dominant-negative manner to reduce Kv7 conductance Cannabidiol acts on Kv7.x activation voltage to offset the effects of Kv7.2mut
Collapse
Affiliation(s)
- Xiaoqin Zhan
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Chris Drummond-Main
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Dylan Greening
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jinjing Yao
- Libin Cardiovascular Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - S W R Chen
- Libin Cardiovascular Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - J P Appendino
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Pediatric Department, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - P Y Billie Au
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Medical Genetics, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Ray W Turner
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
38
|
Hendricks EL, Smith IR, Prates B, Barmaleki F, Liebl FLW. The CD63 homologs, Tsp42Ee and Tsp42Eg, restrict endocytosis and promote neurotransmission through differential regulation of synaptic vesicle pools. Front Cell Neurosci 2022; 16:957232. [PMID: 36072568 PMCID: PMC9441712 DOI: 10.3389/fncel.2022.957232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
The Tetraspanin (Tsp), CD63, is a transmembrane component of late endosomes and facilitates vesicular trafficking through endosomal pathways. Despite being widely expressed in the human brain and localized to late endosomes, CD63's role in regulating endo- and exocytic cycling at the synapse has not been investigated. Synaptic vesicle pools are highly dynamic and disruptions in the mobilization and replenishment of these vesicle pools have adverse neuronal effects. We find that the CD63 homologs, Tsp42Ee and Tsp42Eg, are expressed at the Drosophila neuromuscular junction to regulate synaptic vesicle pools through both shared and unique mechanisms. Tsp42Ee and Tsp42Eg negatively regulate endocytosis and positively regulate neurotransmitter release. Both tsp mutants show impaired locomotion, reduced miniature endplate junctional current frequencies, and increased endocytosis. Expression of human CD63 in Drosophila neurons leads to impaired endocytosis suggesting the role of Tsps in endocytosis is conserved. We further show that Tsps influence the synaptic cytoskeleton and membrane composition by regulating Futsch loop formation and synaptic levels of SCAR and PI(4,5)P2. Finally, Tsp42Ee and Tsp42Eg influence the synaptic localization of several vesicle-associated proteins including Synapsin, Synaptotagmin, and Cysteine String Protein. Together, our results present a novel function for Tsps in the regulation of vesicle pools and provide insight into the molecular mechanisms of Tsp-related synaptic dysfunction.
Collapse
Affiliation(s)
| | | | | | | | - Faith L. W. Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, United States
| |
Collapse
|
39
|
Musella S, Carotenuto L, Iraci N, Baroli G, Ciaglia T, Nappi P, Basilicata MG, Salviati E, Barrese V, Vestuto V, Pignataro G, Pepe G, Sommella E, Di Sarno V, Manfra M, Campiglia P, Gomez-Monterrey I, Bertamino A, Taglialatela M, Ostacolo C, Miceli F. Beyond Retigabine: Design, Synthesis, and Pharmacological Characterization of a Potent and Chemically Stable Neuronal Kv7 Channel Activator with Anticonvulsant Activity. J Med Chem 2022; 65:11340-11364. [PMID: 35972998 PMCID: PMC9421656 DOI: 10.1021/acs.jmedchem.2c00911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
![]()
Neuronal Kv7 channels represent important pharmacological
targets
for hyperexcitability disorders including epilepsy. Retigabine is
the prototype Kv7 activator clinically approved for seizure treatment;
however, severe side effects associated with long-term use have led
to its market discontinuation. Building upon the recently described
cryoEM structure of Kv7.2 complexed with retigabine and on previous
structure–activity relationship studies, a small library of
retigabine analogues has been designed, synthesized, and characterized
for their Kv7 opening ability using both fluorescence- and electrophysiology-based
assays. Among all tested compounds, 60 emerged as a potent
and photochemically stable neuronal Kv7 channel activator. Compared
to retigabine, compound 60 displayed a higher brain/plasma
distribution ratio, a longer elimination half-life, and more potent
and effective anticonvulsant effects in an acute seizure model in
mice. Collectively, these data highlight compound 60 as
a promising lead compound for the development of novel Kv7 activators
for the treatment of hyperexcitability diseases.
Collapse
Affiliation(s)
- Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Lidia Carotenuto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | - Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (CHIBIOFARAM), University of Messina, Viale Ferdinando Stagno d'Alcontres 31, Messina 98166, Italy
| | - Giulia Baroli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Piera Nappi
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | | | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Vincenzo Barrese
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Giuseppe Pignataro
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Michele Manfra
- Department of Science, University of Basilicata, Via dell'Ateneo Lucano 10, Potenza 85100, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Isabel Gomez-Monterrey
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, Naples 80131, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, Fisciano 84084, Salerno, Italy
| | - Maurizio Taglialatela
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, Naples 80131, Italy
| | - Francesco Miceli
- Department of Neuroscience, Reproductive Sciences and Dentistry, University Federico II of Naples, Via Pansini, 5, Naples 80131, Italy
| |
Collapse
|
40
|
Tracy GC, Wilton AR, Rhodes JS, Chung HJ. Heterozygous Deletion of Epilepsy Gene KCNQ2 Has Negligible Effects on Learning and Memory. Front Behav Neurosci 2022; 16:930216. [PMID: 35928789 PMCID: PMC9344800 DOI: 10.3389/fnbeh.2022.930216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal Kv7/Potassium Voltage-Gated Channel Subfamily Q (KCNQ) potassium channels underlie M-current that potently suppresses repetitive and burst firing of action potentials (APs). They are mostly heterotetramers of Kv7.2 and Kv7.3 subunits in the hippocampus and cortex, the brain regions important for cognition and behavior. Underscoring their critical roles in inhibiting neuronal excitability, autosomal dominantly inherited mutations in Potassium Voltage-Gated Channel Subfamily Q Member 2 (KCNQ2) and Potassium Voltage-Gated Channel Subfamily Q Member 3 (KCNQ3) genes are associated with benign familial neonatal epilepsy (BFNE) in which most seizures spontaneously remit within months without cognitive deficits. De novo mutations in KCNQ2 also cause epileptic encephalopathy (EE), which is characterized by persistent seizures that are often drug refractory, neurodevelopmental delay, and intellectual disability. Heterozygous expression of EE variants of KCNQ2 is recently shown to induce spontaneous seizures and cognitive deficit in mice, although it is unclear whether this cognitive deficit is caused directly by Kv7 disruption or by persistent seizures in the developing brain as a consequence of Kv7 disruption. In this study, we examined the role of Kv7 channels in learning and memory by behavioral phenotyping of the KCNQ2+/- mice, which lack a single copy of KCNQ2 but dos not display spontaneous seizures. We found that both KCNQ2+/- and wild-type (WT) mice showed comparable nociception in the tail-flick assay and fear-induced learning and memory during a passive inhibitory avoidance (IA) test and contextual fear conditioning (CFC). Both genotypes displayed similar object location and recognition memory. These findings together provide evidence that heterozygous loss of KCNQ2 has minimal effects on learning or memory in mice in the absence of spontaneous seizures.
Collapse
Affiliation(s)
- Gregory C. Tracy
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Angelina R. Wilton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Justin S. Rhodes
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
41
|
Miceli F, Millevert C, Soldovieri MV, Mosca I, Ambrosino P, Carotenuto L, Schrader D, Lee HK, Riviello J, Hong W, Risen S, Emrick L, Amin H, Ville D, Edery P, de Bellescize J, Michaud V, Van-Gils J, Goizet C, Willemsen MH, Kleefstra T, Møller RS, Bayat A, Devinsky O, Sands T, Korenke GC, Kluger G, Mefford HC, Brilstra E, Lesca G, Milh M, Cooper EC, Taglialatela M, Weckhuysen S. KCNQ2 R144 variants cause neurodevelopmental disability with language impairment and autistic features without neonatal seizures through a gain-of-function mechanism. EBioMedicine 2022; 81:104130. [PMID: 35780567 PMCID: PMC9254340 DOI: 10.1016/j.ebiom.2022.104130] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 01/10/2023] Open
Abstract
Background Prior studies have revealed remarkable phenotypic heterogeneity in KCNQ2-related disorders, correlated with effects on biophysical features of heterologously expressed channels. Here, we assessed phenotypes and functional properties associated with KCNQ2 missense variants R144W, R144Q, and R144G. We also explored in vitro blockade of channels carrying R144Q mutant subunits by amitriptyline. Methods Patients were identified using the RIKEE database and through clinical collaborators. Phenotypes were collected by a standardized questionnaire. Functional and pharmacological properties of variant subunits were analyzed by whole-cell patch-clamp recordings. Findings Detailed clinical information on fifteen patients (14 novel and 1 previously published) was analyzed. All patients had developmental delay with prominent language impairment. R144Q patients were more severely affected than R144W patients. Infantile to childhood onset epilepsy occurred in 40%, while 67% of sleep-EEGs showed sleep-activated epileptiform activity. Ten patients (67%) showed autistic features. Activation gating of homomeric Kv7.2 R144W/Q/G channels was left-shifted, suggesting gain-of-function effects. Amitriptyline blocked channels containing Kv7.2 and Kv7.2 R144Q subunits. Interpretation Patients carrying KCNQ2 R144 gain-of-function variants have developmental delay with prominent language impairment, autistic features, often accompanied by infantile- to childhood-onset epilepsy and EEG sleep-activated epileptiform activity. The absence of neonatal seizures is a robust and important clinical differentiator between KCNQ2 gain-of-function and loss-of-function variants. The Kv7.2/7.3 channel blocker amitriptyline might represent a targeted treatment. Funding Supported by FWO, GSKE, KCNQ2-Cure, Jack Pribaz Foundation, European Joint Programme on Rare Disease 2020, the Italian Ministry for University and Research, the Italian Ministry of Health, the European Commission, the University of Antwerp, NINDS, and Chalk Family Foundation.
Collapse
|
42
|
Inactivation of the Lateral Hypothalamus Attenuates Methamphetamine-Induced Conditioned Place Preference through Regulation of Kcnq3 Expression. Int J Mol Sci 2022; 23:ijms23137305. [PMID: 35806315 PMCID: PMC9266452 DOI: 10.3390/ijms23137305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022] Open
Abstract
Repeated administration of methylamphetamine (MA) induces MA addiction, which is featured by awfully unpleasant physical and emotional experiences after drug use is terminated. Neurophysiological studies show that the lateral hypothalamus (LH) is involved in reward development and addictive behaviors. Here, we show that repeated administration of MA activates the expression of c-Fos in LH neurons responding to conditioned place preference (CPP). Chemogenetic inhibition of the LH can disrupt the addiction behavior, demonstrating that the LH plays an important role in MA-induced reward processing. Critically, MA remodels the neurons of LH synaptic plasticity, increases intracellular calcium level, and enhances spontaneous current and evoked potentials of neurons compared to the saline group. Furthermore, overexpression of the potassium voltage-gated channel subfamily Q member 3 (Kcnq3) expression can reverse the CPP score and alleviate the occurrence of addictive behaviors. Together, these results unravel a new neurobiological mechanism underlying the MA-induced addiction in the lateral hypothalamus, which could pave the way toward new and effective interventions for this addiction disease.
Collapse
|
43
|
Soldovieri MV, Taglialatela M. The long and winding road to personalized medicine in KCNMA1-linked channelopathies revealed by novel variants associated with the Liang-Wang syndrome. Acta Physiol (Oxf) 2022; 235:e13854. [PMID: 35730691 DOI: 10.1111/apha.13854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Maria Virginia Soldovieri
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | | |
Collapse
|
44
|
Frampton DJA, Choudhury K, Nikesjö J, Delemotte L, Liin SI. Subtype-specific responses of hKv7.4 and hKv7.5 channels to polyunsaturated fatty acids reveal an unconventional modulatory site and mechanism. eLife 2022; 11:77672. [PMID: 35642964 PMCID: PMC9159753 DOI: 10.7554/elife.77672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
The KV7.4 and KV7.5 subtypes of voltage-gated potassium channels play a role in important physiological processes such as sound amplification in the cochlea and adjusting vascular smooth muscle tone. Therefore, the mechanisms that regulate KV7.4 and KV7.5 channel function are of interest. Here, we study the effect of polyunsaturated fatty acids (PUFAs) on human KV7.4 and KV7.5 channels expressed in Xenopus oocytes. We report that PUFAs facilitate activation of hKV7.5 by shifting the V50 of the conductance versus voltage (G(V)) curve toward more negative voltages. This response depends on the head group charge, as an uncharged PUFA analogue has no effect and a positively charged PUFA analogue induces positive V50 shifts. In contrast, PUFAs inhibit activation of hKV7.4 by shifting V50 toward more positive voltages. No effect on V50 of hKV7.4 is observed by an uncharged or a positively charged PUFA analogue. Thus, the hKV7.5 channel's response to PUFAs is analogous to the one previously observed in hKV7.1-7.3 channels, whereas the hKV7.4 channel response is opposite, revealing subtype-specific responses to PUFAs. We identify a unique inner PUFA interaction site in the voltage-sensing domain of hKV7.4 underlying the PUFA response, revealing an unconventional mechanism of modulation of hKV7.4 by PUFAs.
Collapse
Affiliation(s)
- Damon J A Frampton
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Johan Nikesjö
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Sara I Liin
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
45
|
Gain of function due to increased opening probability by two KCNQ5 pore variants causing developmental and epileptic encephalopathy. Proc Natl Acad Sci U S A 2022; 119:e2116887119. [PMID: 35377796 PMCID: PMC9169635 DOI: 10.1073/pnas.2116887119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Variants in genes encoding neuronally expressed potassium channel subunits are frequent causes of developmental and epileptic encephalopathies (DEEs). Characterization of their functional consequences is critical to confirm diagnosis, assess prognosis, and implement personalized treatments. In the present work, we describe two patients carrying variants in KCNQ5, a gene very recently and rarely found involved in DEEs, and reveal that they both cause remarkable gain-of-function consequences on channel activity. A PIP2-independent increase in open probability, without effects on membrane abundance or single-channel conductance, was responsible for the observed mutation-induced functional changes, thus revealing a pathomolecular disease mechanism for DEEs. Developmental and epileptic encephalopathies (DEEs) are neurodevelopmental diseases characterized by refractory epilepsy, distinct electroencephalographic and neuroradiological features, and various degrees of developmental delay. Mutations in KCNQ2, KCNQ3, and, more rarely, KCNQ5 genes encoding voltage-gated potassium channel subunits variably contributing to excitability control of specific neuronal populations at distinct developmental stages have been associated to DEEs. In the present work, the clinical features of two DEE patients carrying de novo KCNQ5 variants affecting the same residue in the pore region of the Kv7.5 subunit (G347S/A) are described. The in vitro functional properties of channels incorporating these variants were investigated with electrophysiological and biochemical techniques to highlight pathophysiological disease mechanisms. Currents carried by Kv7.5 G347 S/A channels displayed: 1) large (>10 times) increases in maximal current density, 2) the occurrence of a voltage-independent component, 3) slower deactivation kinetics, and 4) hyperpolarization shift in activation. All these functional features are consistent with a gain-of-function (GoF) pathogenetic mechanism. Similar functional changes were also observed when the same variants were introduced at the corresponding position in Kv7.2 subunits. Nonstationary noise analysis revealed that GoF effects observed for both Kv7.2 and Kv7.5 variants were mainly attributable to an increase in single-channel open probability, without changes in membrane abundance or single-channel conductance. The mutation-induced increase in channel opening probability was insensitive to manipulation of membrane levels of the critical Kv7 channel regulator PIP2. These results reveal a pathophysiological mechanism for KCNQ5-related DEEs, which might be exploited to implement personalized treatments.
Collapse
|
46
|
Abstract
KCNQ2 and KCNQ3 channels are associated with multiple neurodevelopmental disorders and are also therapeutic targets for neurological and neuropsychiatric diseases. For more than two decades, it has been thought that most KCNQ channels in the brain are either KCNQ2/3 or KCNQ3/5 heteromers. Here, we investigated the potential heteromeric compositions of KCNQ2-containing channels. We applied split-intein protein trans-splicing to form KCNQ2/5 tandems and coexpressed these with and without KCNQ3. Unexpectedly, we found that KCNQ2/5 tandems form functional channels independent of KCNQ3 in heterologous cells. Using mass spectrometry, we went on to demonstrate that KCNQ2 associates with KCNQ5 in native channels in the brain, even in the absence of KCNQ3. Additionally, our functional heterologous expression data are consistent with the formation of KCNQ2/3/5 heteromers. Thus, the composition of KCNQ channels is more diverse than has been previously recognized, necessitating a re-examination of the genotype/phenotype relationship of KCNQ2 pathogenic variants.
Collapse
|
47
|
Xiong J, Chen S, Chen B, Zhang W, Chen C, Deng X, He F, Zhang C, Yang L, Wang Y, Peng J, Yin F. A novel KCNQ2 missense variant in non-syndromic intellectual disability causes mild gain-of-function of Kv7.2 channel. Clin Chim Acta 2022; 530:74-80. [PMID: 35247435 DOI: 10.1016/j.cca.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Heterozygous variants of KCNQ2 can cause KCNQ2 associated neurodevelopmental disorder, mainly are benign (familial) neonatal or infantile epilepsy (B(F)NE or B(F)IE) and early-onset epileptic encephalopathy (EOEE). Moreover, some intermediate phenotypes, including intellectual disability (ID), and myokymia are related to the gene. METHODS We collected a non-syndromic ID male patient with a novel KCNQ2 missense variant. Whole cell electrophysiology, western blotting, and immunofluorescence were adopted to analyze the variant's functional alterations. RESULTS The patient presented with global developmental delay since his infancy. He still had profound ID but did not have epilepsy at the adolescence. The de novo KCNQ2 variant p.R75C (NM_172107) in the NH2 domain identified here showed a slightly hyperpolarized shift of activation curves and larger current density in homomeric configurations, which could be abolished in co-expression with Kv7.2 or Kv7.3 wild-type. Western blotting and immunocytochemistry supported that the expression of variant p.R75C is lower than the Kv7.2 wild-type. The findings indicated variant p.R75C cause mild gain-of-function (GOF) of Kv7.2 channel. CONCLUSIONS We report a non-syndromic ID patient with a KCNQ2 mild GOF variant, adding evidence for this rare clinical phenotype in the disorder. We propose that individuals with KCNQ2 GOF variants are prone to have cognitive impairments.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Shimeng Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Wen Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Chen Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Xiaolu Deng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ciliu Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ying Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China; Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China.
| |
Collapse
|
48
|
Abdullaeva OS, Sahalianov I, Silverå Ejneby M, Jakešová M, Zozoulenko I, Liin SI, Głowacki ED. Faradaic Pixels for Precise Hydrogen Peroxide Delivery to Control M-Type Voltage-Gated Potassium Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103132. [PMID: 34825522 PMCID: PMC8787424 DOI: 10.1002/advs.202103132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/28/2021] [Indexed: 06/13/2023]
Abstract
H2 O2 plays a significant role in a range of physiological processes where it performs vital tasks in redox signaling. The sensitivity of many biological pathways to H2 O2 opens up a unique direction in the development of bioelectronics devices to control levels of reactive-oxygen species (ROS). Here a microfabricated ROS modulation device that relies on controlled faradaic reactions is presented. A concentric pixel arrangement of a peroxide-evolving cathode surrounded by an anode ring which decomposes the peroxide, resulting in localized peroxide delivery is reported. The conducting polymer (poly(3,4-ethylenedioxythiophene) (PEDOT), is exploited as the cathode. PEDOT selectively catalyzes the oxygen reduction reaction resulting in the production of hydrogen peroxide (H2 O2 ). Using electrochemical and optical assays, combined with modeling, the performance of the devices is benchmarked. The concentric pixels generate tunable gradients of peroxide and oxygen concentrations. The faradaic devices are prototyped by modulating human H2 O2 -sensitive Kv7.2/7.3 (M-type) channels expressed in a single-cell model (Xenopus laevis oocytes). The Kv7 ion channel family is responsible for regulating neuronal excitability in the heart, brain, and smooth muscles, making it an ideal platform for faradaic ROS stimulation. The results demonstrate the potential of PEDOT to act as an H2 O2 delivery system, paving the way to ROS-based organic bioelectronics.
Collapse
Affiliation(s)
- Oliya S. Abdullaeva
- Laboratory of Organic ElectronicsITN Campus NorrköpingLinköping UniversityNorrköpingSE‐60174Sweden
- Wallenberg Center for Molecular MedicineLinköping UniversityLinköpingSE‐58185Sweden
| | - Ihor Sahalianov
- Laboratory of Organic ElectronicsITN Campus NorrköpingLinköping UniversityNorrköpingSE‐60174Sweden
| | - Malin Silverå Ejneby
- Laboratory of Organic ElectronicsITN Campus NorrköpingLinköping UniversityNorrköpingSE‐60174Sweden
- Wallenberg Center for Molecular MedicineLinköping UniversityLinköpingSE‐58185Sweden
| | - Marie Jakešová
- Bioelectronics Materials and Devices LabCentral European Institute of TechnologyBrno University of TechnologyPurkyňova 123Brno61200Czech Republic
| | - Igor Zozoulenko
- Laboratory of Organic ElectronicsITN Campus NorrköpingLinköping UniversityNorrköpingSE‐60174Sweden
| | - Sara I. Liin
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSE‐58185Sweden
| | - Eric Daniel Głowacki
- Laboratory of Organic ElectronicsITN Campus NorrköpingLinköping UniversityNorrköpingSE‐60174Sweden
- Wallenberg Center for Molecular MedicineLinköping UniversityLinköpingSE‐58185Sweden
- Bioelectronics Materials and Devices LabCentral European Institute of TechnologyBrno University of TechnologyPurkyňova 123Brno61200Czech Republic
| |
Collapse
|
49
|
Martin-Batista E, Manville RW, Rivero-Pérez B, Bartolomé-Martín D, Alvarez de la Rosa D, Abbott GW, Giraldez T. Activation of SGK1.1 Upregulates the M-current in the Presence of Epilepsy Mutations. Front Mol Neurosci 2021; 14:798261. [PMID: 34899186 PMCID: PMC8662703 DOI: 10.3389/fnmol.2021.798261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
In the central nervous system, the M-current plays a critical role in regulating subthreshold electrical excitability of neurons, determining their firing properties and responsiveness to synaptic input. The M-channel is mainly formed by subunits Kv7.2 and Kv7.3 that co-assemble to form a heterotetrametric channel. Mutations in Kv7.2 and Kv7.3 are associated with hyperexcitability phenotypes including benign familial neonatal epilepsy (BFNE) and neonatal epileptic encephalopathy (NEE). SGK1.1, the neuronal isoform of the serum and glucocorticoids-regulated kinase 1 (SGK1), increases M-current density in neurons, leading to reduced excitability and protection against seizures. Herein, using two-electrode voltage clamp on Xenopus laevis oocytes, we demonstrate that SGK1.1 selectively activates heteromeric Kv7 subunit combinations underlying the M-current. Importantly, activated SGK1.1 increases M-channel activity in the presence of two different epilepsy mutations found in Kv7.2, R207W and A306T. In addition, proximity ligation assays in the N2a cell line allowed us to address the effect of these mutations on Kv7-SGK1.1-Nedd4 molecular associations, a proposed pathway underlying augmentation of M-channel activity by SGK1.1.
Collapse
Affiliation(s)
- Elva Martin-Batista
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Belinda Rivero-Pérez
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - David Bartolomé-Martín
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Teresa Giraldez
- Departamento de Ciencias Medicas Basicas and Instituto de Tecnologias Biomedicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
50
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|